Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Cyriac Kandoth is active.

Publication


Featured researches published by Cyriac Kandoth.


Leukemia | 2011

Recurrent DNMT3A mutations in patients with myelodysplastic syndromes

Matthew J. Walter; Li Ding; Dong Shen; Jin Shao; Marcus Grillot; Michael D. McLellan; Robert S. Fulton; Heather K. Schmidt; Joelle Kalicki-Veizer; Michelle O'Laughlin; Cyriac Kandoth; Jack Baty; Peter Westervelt; John F. DiPersio; Elaine R. Mardis; Richard Wilson; Timothy J. Ley; Timothy A. Graubert

Alterations in DNA methylation have been implicated in the pathogenesis of myelodysplastic syndromes (MDS), although the underlying mechanism remains largely unknown. Methylation of CpG dinucleotides is mediated by DNA methyltransferases, including DNMT1, DNMT3A and DNMT3B. DNMT3A mutations have recently been reported in patients with de novo acute myeloid leukemia (AML), providing a rationale for examining the status of DNMT3A in MDS samples. In this study, we report the frequency of DNMT3A mutations in patients with de novo MDS, and their association with secondary AML. We sequenced all coding exons of DNMT3A using DNA from bone marrow and paired normal cells from 150 patients with MDS and identified 13 heterozygous mutations with predicted translational consequences in 12/150 patients (8.0%). Amino acid R882, located in the methyltransferase domain of DNMT3A, was the most common mutation site, accounting for 4/13 mutations. DNMT3A mutations were expressed in the majority of cells in all tested mutant samples regardless of myeloblast counts, suggesting that DNMT3A mutations occur early in the course of MDS. Patients with DNMT3A mutations had worse overall survival compared with patients without DNMT3A mutations (P=0.005) and more rapid progression to AML (P=0.007), suggesting that DNMT3A mutation status may have prognostic value in de novo MDS.


Nature Genetics | 2013

Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas

Junyuan Zhang; Gang Wu; Cp Miller; Ruth G. Tatevossian; James Dalton; Bo Tang; Wilda Orisme; Chandanamali Punchihewa; Michael W. Parker; Ibrahim Qaddoumi; F.A. Boop; Charles Lu; Cyriac Kandoth; Li Ding; Ryan Lee; Robert Huether; Xian Chen; Erin Hedlund; Panduka Nagahawatte; Michael Rusch; Kristy Boggs; Jinjun Cheng; Jared Becksfort; Jing Ma; Guangchun Song; Yongjin Li; Lei Wei; Jioajiao Wang; Sheila A. Shurtleff; John Easton

The most common pediatric brain tumors are low-grade gliomas (LGGs). We used whole-genome sequencing to identify multiple new genetic alterations involving BRAF, RAF1, FGFR1, MYB, MYBL1 and genes with histone-related functions, including H3F3A and ATRX, in 39 LGGs and low-grade glioneuronal tumors (LGGNTs). Only a single non-silent somatic alteration was detected in 24 of 39 (62%) tumors. Intragenic duplications of the portion of FGFR1 encoding the tyrosine kinase domain (TKD) and rearrangements of MYB were recurrent and mutually exclusive in 53% of grade II diffuse LGGs. Transplantation of Trp53-null neonatal astrocytes expressing FGFR1 with the duplication involving the TKD into the brains of nude mice generated high-grade astrocytomas with short latency and 100% penetrance. FGFR1 with the duplication induced FGFR1 autophosphorylation and upregulation of the MAPK/ERK and PI3K pathways, which could be blocked by specific inhibitors. Focusing on the therapeutically challenging diffuse LGGs, our study of 151 tumors has discovered genetic alterations and potential therapeutic targets across the entire range of pediatric LGGs and LGGNTs.


Leukemia | 2013

Clonal diversity of recurrently mutated genes in myelodysplastic syndromes

Matthew J. Walter; Dong Shen; Jin Shao; Li Ding; Brian S. White; Cyriac Kandoth; Christopher A. Miller; Beifang Niu; McLellan; Nathan D. Dees; Robert S. Fulton; K Elliot; Simon Heath; Marcus Grillot; Peter Westervelt; Daniel C. Link; John F. DiPersio; Elaine R. Mardis; Timothy J. Ley; Richard Wilson; Timothy A. Graubert

Recent studies suggest that most cases of myelodysplastic syndrome (MDS) are clonally heterogeneous, with a founding clone and multiple subclones. It is not known whether specific gene mutations typically occur in founding clones or subclones. We screened a panel of 94 candidate genes in a cohort of 157 patients with MDS or secondary acute myeloid leukemia (sAML). This included 150 cases with samples obtained at MDS diagnosis and 15 cases with samples obtained at sAML transformation (8 were also analyzed at the MDS stage). We performed whole-genome sequencing (WGS) to define the clonal architecture in eight sAML genomes and identified the range of variant allele frequencies (VAFs) for founding clone mutations. At least one mutation or cytogenetic abnormality was detected in 83% of the 150 MDS patients and 17 genes were significantly mutated (false discovery rate ⩽0.05). Individual genes and patient samples displayed a wide range of VAFs for recurrently mutated genes, indicating that no single gene is exclusively mutated in the founding clone. The VAFs of recurrently mutated genes did not fully recapitulate the clonal architecture defined by WGS, suggesting that comprehensive sequencing may be required to accurately assess the clonal status of recurrently mutated genes in MDS.


Nature Biotechnology | 2016

Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity

Matthew T. Chang; Saurabh Asthana; Sizhi Paul Gao; Byron H. Lee; Jocelyn S. Chapman; Cyriac Kandoth; Jianjiong Gao; Nicholas D. Socci; David B. Solit; Adam B. Olshen; Nikolaus Schultz; Barry S. Taylor

Mutational hotspots indicate selective pressure across a population of tumor samples, but their prevalence within and across cancer types is incompletely characterized. An approach to detect significantly mutated residues, rather than methods that identify recurrently mutated genes, may uncover new biologically and therapeutically relevant driver mutations. Here, we developed a statistical algorithm to identify recurrently mutated residues in tumor samples. We applied the algorithm to 11,119 human tumors, spanning 41 cancer types, and identified 470 somatic substitution hotspots in 275 genes. We find that half of all human tumors possess one or more mutational hotspots with widespread lineage-, position- and mutant allele–specific differences, many of which are likely functional. In total, 243 hotspots were novel and appeared to affect a broad spectrum of molecular function, including hotspots at paralogous residues of Ras-related small GTPases RAC1 and RRAS2. Redefining hotspots at mutant amino acid resolution will help elucidate the allele-specific differences in their function and could have important therapeutic implications.


Cell | 2018

Tumor Evolution and Drug Response in Patient-Derived Organoid Models of Bladder Cancer

Suk Hyung Lee; Wenhuo Hu; Justin T. Matulay; Mark V. Silva; Tomasz Owczarek; Kwanghee Kim; Chee Wai Chua; LaMont Barlow; Cyriac Kandoth; Alanna B. Williams; Sarah K. Bergren; Eugene J. Pietzak; Christopher B. Anderson; Mitchell C. Benson; Jonathan A. Coleman; Barry S. Taylor; Cory Abate-Shen; James M. McKiernan; Hikmat Al-Ahmadie; David B. Solit; Michael M. Shen

Bladder cancer is the fifth most prevalent cancer in the U.S., yet is understudied, and few laboratory models exist that reflect the biology of the human disease. Here, we describe a biobank of patient-derived organoid lines that recapitulates the histopathological and molecular diversity of human bladder cancer. Organoid lines can be established efficiently from patient biopsies acquired before and after disease recurrence and are interconvertible with orthotopic xenografts. Notably, organoid lines often retain parental tumor heterogeneity and exhibit a spectrum of genomic changes that are consistent with tumor evolution in culture. Analyses of drug response using bladder tumor organoids show partial correlations with mutational profiles, as well as changes associated with treatment resistance, and specific responses can be validated using xenografts inxa0vivo. Our studies indicate that patient-derived bladder tumor organoids represent a faithful model system for studying tumor evolution and treatment response in the context of precision cancer medicine.


Cancer Discovery | 2017

Accelerating Discovery of Functional Mutant Alleles in Cancer

Matthew T. Chang; Tripti Shrestha Bhattarai; Alison M. Schram; Craig M. Bielski; Mark T.A. Donoghue; Philip Jonsson; Debyani Chakravarty; Sarah Phillips; Cyriac Kandoth; Alexander Penson; Alexander N. Gorelick; Tambudzai Shamu; Swati Patel; Christopher C. Harris; Jianjiong Gao; Selcuk Onur Sumer; Ritika Kundra; Pedram Razavi; Bob T. Li; Dalicia Reales; Nicholas D. Socci; Gowtham Jayakumaran; Ahmet Zehir; Ryma Benayed; Maria E. Arcila; Sarat Chandarlapaty; Marc Ladanyi; Nikolaus Schultz; José Baselga; Michael F. Berger

Most mutations in cancer are rare, which complicates the identification of therapeutically significant mutations and thus limits the clinical impact of genomic profiling in patients with cancer. Here, we analyzed 24,592 cancers including 10,336 prospectively sequenced patients with advanced disease to identify mutant residues arising more frequently than expected in the absence of selection. We identified 1,165 statistically significant hotspot mutations of which 80% arose in 1 in 1,000 or fewer patients. Of 55 recurrent in-frame indels, we validated that novel AKT1 duplications induced pathway hyperactivation and conferred AKT inhibitor sensitivity. Cancer genes exhibit different rates of hotspot discovery with increasing sample size, with few approaching saturation. Consequently, 26% of all hotspots in therapeutically actionable oncogenes were novel. Upon matching a subset of affected patients directly to molecularly targeted therapy, we observed radiographic and clinical responses. Population-scale mutant allele discovery illustrates how the identification of driver mutations in cancer is far from complete.Significance: Our systematic computational, experimental, and clinical analysis of hotspot mutations in approximately 25,000 human cancers demonstrates that the long right tail of biologically and therapeutically significant mutant alleles is still incompletely characterized. Sharing prospective genomic data will accelerate hotspot identification, thereby expanding the reach of precision oncology in patients with cancer. Cancer Discov; 8(2); 174-83. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 127.


Cancer Research | 2015

Abstract S2-04: Comprehensive molecular characterization of invasive lobular breast tumors

Giovanni Ciriello; Michael L. Gatza; Katherine A. Hoadley; Hailei Zhang; Suhn Kyong Rhie; Reanne Bowlby; Matthew D. Wilkerson; Cyriac Kandoth; Michael D. McLellan; Andrew D. Cherniack; Peter W. Laird; Chris Sander; Tari A. King; Charles M. Perou

Invasive lobular breast cancer (ILC) is the second most common histological subtype of breast cancer accounting for 10-15% of invasive breast tumors. ILC is typically ER+ and beyond the known mutation and/or loss of E-cadherin function, which contributes to a highly discohesive morphology, little is known about the additional mechanisms driving ILC tumorigenesis, or alterations that differentiate ILC from invasive ductal carcinomas (IDC). Methods A dataset of 817 breast tumors from the TCGA Project, including 490 IDC, 127 ILC and 88 samples with a mixed IDC-ILC histology, were profiled on six genomic platforms to develop a comprehensive atlas of mutational, epigenetic, transcriptional and proteomic data. Integrative genomic analyses, both supervised and unsupervised, of ILC tumors and across histological subtypes were performed to identify genomic drivers of ILC oncogenesis. Results Comprehensive multi-platform analyses identified distinct molecular events associated with ILC tumors. As expected, lack of E-cadherin protein, as determined by Reverse Phase Protein Array (RPPA), and CDH1 mRNA expression was uniformly observed in ILC cases associated with distinct alterations targeting CDH1. In addition to previously reported CDH1 and PIK3CA mutations, we identified a number of novel ILC-enriched recurrent mutations targeting PTEN, RUNX1, TBX3, and FOXA1. An increased incidence of PTEN inactivating events, both mutations and copy number changes, were identified in ILC (13%) compared to IDC ER+ (7%), which corresponded with altered PTEN protein expression. These alterations were largely mutually exclusive with PIK3CA mutations and correlate with increased Akt activation as evident by increased Akt phosphorylation (pS473 and pT308), thus identifying a potential therapeutic opportunity for ILC patients. GATA3 signaling, which regulates epithelial cell differentiation, is frequently altered in luminal/ER+ breast cancers. Our analyses determined GATA3 mutations are more frequent in IDC luminal tumors as compared to ILC (19 % vs 5%). ILC luminal tumors show significantly lower GATA3 protein expression, but a higher frequency of mutations in FOXA1 (9% vs 2% in Luminal IDC), a transcription factor required to promote ER transcriptional programs. Within ILC tumors, FOXA1 mutations were found to cluster into a specific region of the Forkhead (FK) DNA binding domain. A broader analysis of FOXA1 mutations in breast and prostate cancer confirm two specific hotspots in the FK domain and the C-terminal transactivation domain. Interestingly, these mutational classes are associated with distinct transcriptional changes suggesting different functional effects. Finally, mRNA-seq analyses identified three robust molecular subclasses that are characterized by distinct genetic, genomic and proteomic patterns, including an increased immune-related group (Class 2), as well as differences in prognosis. Conclusions In this study, we developed a comprehensive atlas of genomic alterations that reveals key molecular differences differentiating ILC (FOXA1) from IDC (GATA3) tumorigenesis, a potential therapeutic target for ILC (Akt), and novel ILC subclasses based on underlying biological events. These findings provide further insight into the molecular heterogeneity of ER+ breast cancer. Citation Format: Giovanni Ciriello, Michael L Gatza, Katherine A Hoadley, Hailei Zhang, Suhn K Rhie, Reanne Bowlby, Matthew D Wilkerson, Cyriac Kandoth, Michael McLellan, Andrew Cherniack, Peter W Laird, Chris Sander, Tari A King, Charles M Perou. Comprehensive molecular characterization of invasive lobular breast tumors [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr S2-04.


pacific symposium on biocomputing | 2014

Integrative genome-wide analysis of the determinants of RNA splicing in kidney renal clear cell carcinoma.

Kjong-Van Lehmann; André Kahles; Cyriac Kandoth; William Lee; Nikolaus Schultz; Oliver Stegle; Gunnar Rätsch

We present a genome-wide analysis of splicing patterns of 282 kidney renal clear cell carcinoma patients in which we integrate data from whole-exome sequencing of tumor and normal samples, RNA-seq and copy number variation. We proposed a scoring mechanism to compare splicing patterns in tumor samples to normal samples in order to rank and detect tumor-specific isoforms that have a potential for new biomarkers. We identified a subset of genes that show introns only observable in tumor but not in normal samples, ENCODE and GEUVADIS samples. In order to improve our understanding of the underlying genetic mechanisms of splicing variation we performed a large-scale association analysis to find links between somatic or germline variants with alternative splicing events. We identified 915 cis- and trans-splicing quantitative trait loci (sQTL) associated with changes in splicing patterns. Some of these sQTL have previously been associated with being susceptibility loci for cancer and other diseases. Our analysis also allowed us to identify the function of several COSMIC variants showing significant association with changes in alternative splicing. This demonstrates the potential significance of variants affecting alternative splicing events and yields insights into the mechanisms related to an array of disease phenotypes.


Genome Medicine | 2018

Integrative omics analyses broaden treatment targets in human cancer

Sohini Sengupta; Sam Q. Sun; Kuan-lin Huang; Clara Oh; Matthew Bailey; Rajees Varghese; Matthew A. Wyczalkowski; Jie Ning; Piyush Tripathi; Joshua F. McMichael; Kimberly J. Johnson; Cyriac Kandoth; John S. Welch; Cynthia X. Ma; Michael C. Wendl; Samuel H. Payne; David Fenyö; R. Reid Townsend; John F. DiPersio; Feng Chen; Li Ding

BackgroundAlthough large-scale, next-generation sequencing (NGS) studies of cancers hold promise for enabling precision oncology, challenges remain in integrating NGS with clinically validated biomarkers.MethodsTo overcome such challenges, we utilized the Database of Evidence for Precision Oncology (DEPO) to link druggability to genomic, transcriptomic, and proteomic biomarkers. Using a pan-cancer cohort of 6570 tumors, we identified tumors with potentially druggable biomarkers consisting of drug-associated mutations, mRNA expression outliers, and protein/phosphoprotein expression outliers identified by DEPO.ResultsWithin the pan-cancer cohort of 6570 tumors, we found that 3% are druggable based on FDA-approved drug-mutation interactions in specific cancer types. However, mRNA/phosphoprotein/protein expression outliers and drug repurposing across cancer types suggest potential druggability in up to 16% of tumors. The percentage of potential drug-associated tumors can increase to 48% if we consider preclinical evidence. Further, our analyses showed co-occurring potentially druggable multi-omics alterations in 32% of tumors, indicating a role for individualized combinational therapy, with evidence supporting mTOR/PI3K/ESR1 co-inhibition and BRAF/AKT co-inhibition in 1.6 and 0.8% of tumors, respectively. We experimentally validated a subset of putative druggable mutations in BRAF identified by a protein structure-based computational tool. Finally, analysis of a large-scale drug screening dataset lent further evidence supporting repurposing of drugs across cancer types and the use of expression outliers for inferring druggability.ConclusionsOur results suggest that an integrated analysis platform can nominate multi-omics alterations as biomarkers of druggability and aid ongoing efforts to bring precision oncology to patients.


Cancer Cell | 2018

The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers

Pedram Razavi; Matthew T. Chang; Guotai Xu; Chaitanya Bandlamudi; Dara S. Ross; Neil Vasan; Yanyan Cai; Craig M. Bielski; Mark T.A. Donoghue; Philip Jonsson; Alexander Penson; Ronglai Shen; Fresia Pareja; Ritika Kundra; Sumit Middha; Michael L. Cheng; Ahmet Zehir; Cyriac Kandoth; Ruchi Patel; Kety Huberman; Lillian Mary Smyth; Komal Jhaveri; Shanu Modi; Tiffany A. Traina; Chau Dang; Wen Zhang; Britta Weigelt; Bob T. Li; Marc Ladanyi; David M. Hyman

We integrated the genomic sequencing of 1,918 breast cancers, including 1,501 hormone receptor-positive tumors, with detailed clinical information and treatment outcomes. In 692 tumors previously exposed to hormonal therapy, we identified an increased number of alterations in genes involved in the mitogen-activated protein kinase (MAPK) pathway and in the estrogen receptor transcriptional machinery. Activating ERBB2 mutations and NF1 loss-of-function mutations were more than twice as common in endocrine resistant tumors. Alterations in other MAPK pathway genes (EGFR, KRAS, among others) and estrogen receptor transcriptional regulators (MYC, CTCF, FOXA1, and TBX3) were also enriched. Altogether, these alterations were present in 22% of tumors, mutually exclusive with ESR1 mutations, and associated with a shorter duration of response to subsequent hormonal therapies.

Collaboration


Dive into the Cyriac Kandoth's collaboration.

Top Co-Authors

Avatar

Barry S. Taylor

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David B. Solit

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Chang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nikolaus Schultz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nicholas D. Socci

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jianjiong Gao

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Li Ding

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ahmet Zehir

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David M. Hyman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge